EP1942872A2 - Esters de carboxyalkylcellulose pour administration d'agents pharmaceutiquement actifs peu solubles - Google Patents

Esters de carboxyalkylcellulose pour administration d'agents pharmaceutiquement actifs peu solubles

Info

Publication number
EP1942872A2
EP1942872A2 EP06827500A EP06827500A EP1942872A2 EP 1942872 A2 EP1942872 A2 EP 1942872A2 EP 06827500 A EP06827500 A EP 06827500A EP 06827500 A EP06827500 A EP 06827500A EP 1942872 A2 EP1942872 A2 EP 1942872A2
Authority
EP
European Patent Office
Prior art keywords
composition according
degree
ranges
alkyl
substitution per
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06827500A
Other languages
German (de)
English (en)
Inventor
Michael Charles Shelton
Jessica Dee Posey-Dowty
Kevin Joseph Edgar
Larry Ronnie Lingerfelt, Jr.
Sandra Klein
Shane Kipley Kirk
Jennifer Joy Dressman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eastman Chemical Co
Original Assignee
Eastman Chemical Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eastman Chemical Co filed Critical Eastman Chemical Co
Publication of EP1942872A2 publication Critical patent/EP1942872A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/717Celluloses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions

Definitions

  • compositions comprising carboxyalkylcellulose esters for delivery of poorly soluble pharmaceutically active substances, e.g., having low solubility in a medium. Also disclosed are methods for making such pharmaceutical compositions and methods of administering the compositions.
  • Solubility and the dissolution profile of a drug in media such as water, aqueous buffers (e.g. simulated gastric fluid (with or without pesin) and simulated intestinal fluid with or without pancreatin)) or in biorelevant media are parameters often used to assess the bioavailability of a drug substance.
  • a drug formulation enters a physiological environment where the drug dissolves and remains in solution.
  • some drug substances fail to dissolve, or may precipitate over time (sometimes due to changes in pH).
  • the pharmaceutical industry is interested in the fate of the drug formulation following introduction to the physiological environment.
  • Drug solubility has been a common limitation in the development of new drug formulations. More than a third of the drugs listed in the United States Pharmacopoeia are poorly soluble or are insoluble in water. (S. Pace et al, Pharm. Tech., pp. 116-132, March, 1999.) Additionally, it is well known that for many drugs the rate-limiting step for the absorption within the gastrointestinal tract is its dissolution. (D.Q.M. Craig et al., Int. J. Pharm., Vol. 179, pp. 179-207, 1999.) To enhance the dissolution rate of poorly water soluble drug and to increase their bioavailability, several techniques have been developed, such as formulation strategies including the formation of solid dispersions. However, such formulations can often be thermodynamically unstable and/or cause undesired side effects.
  • compositions that improve the solubility and or dissolution of poorly water soluble pharmaceutically active agents.
  • FIG. 1 shows carbamazepine and carbamazepine solid dispersions dissolution profiles
  • FIG. 2 shows glyburide and glyburide solid dispersions dissolution profiles
  • FIG. 3 shows glyburide solid dispersions dissolution profiles
  • FIG. 4 shows a comparison of CMCAB/glyburide solid dispersion dissolution profiles (prepared by co-evaporation);
  • FIG. 5 shows a comparison of HPMCAS/glyburide solid dispersion profiles with those of CMCAB/glyburide solid dispersions
  • FIG. 6 shows a comparison of HPMCAS/glyburide solid dispersion dissolution profiles with those of CMCAB/glyburide solid dispersions
  • FIG. 7 shows a dissolution profile of griseofulvin solid dispersions
  • FIG. 8 shows a comparison of CMCAB/griseofulvin solid dispersions dissolution profiles (% released);
  • FIG. 9 shows a comparison of CMCAB/griseofulvin solid dispersions dissolution profiles (mg released);
  • FIG. 10 shows a comparison of CMCAB/griseofulvin solid dispersion profiles with those of PVP/griseofulvin (% released);
  • FIG. 11 shows a comparison of CMCAB/griseofulvin solid dispersions dissolution profiles with those of PVP/griseofulvin solid dispersions (mg released);
  • FIG. 12 shows a comparison of CMCAB/griseofulvin solid dispersions dissolution profiles with those of HPMCAS/griseofulvin solid dispersions (% released);
  • FIG. 13 shows a comparison of CMCAB/griseofulvin solid dispersions dissolution profiles with those of HPMCAS/griseofulvin solid dispersions (mg released);
  • FIG. 14 shows griseofulvin, griseofulvin/CMCAB, and griseofulvin/CMCAB/surfactant solid dispersion dissolution profile comparisons
  • FIG. 15 shows the impact of TPGS on % crystallinity of ibuprofen/CMCAB solid dispersions (D-Optimal Mixture DOE Results).
  • compositions comprising carboxyalkylcellulose esters for administering pharmaceutically active agents to a subject.
  • One embodiment disclosed herein provides a pharmaceutical composition comprising: at least one pharmaceutically active agent having low solubility in a medium, and at least one carboxyalkylcellulose ester comprising an anhydroglucose repeat unit having the structure:
  • R 1 -R 6 are each independently selected from -OH, -OC(O)(alkyl), and
  • - a degree of substitution per anhydroglucose of -OH ranges from 0.1 to 0.7
  • - a degree of substitution per anhydroglucose of -OC(O)(alkyl) ranges from 0.1 to 2.7
  • “Degree of substitution” as used herein refers to a number of substituents per anhydroglucose. A theoretical maximum degree of substitution is 3 is assumed unless stated otherwise as in HS-CMC (high solids carboxymethylcellulose) esters or low molecular weight CMC esters, which can have a maximum degree of substitution per anhydroglucose unit of greater than 3.0.
  • the pharmaceutically acceptable salts include pharmaceutically acceptable salts of -OH and -O(CH 2 ) X C(O)OH having the structure O ' A + and -O(CH 2 ) X C(O)O " A + , respectively, wherein A + is a counterion.
  • Exemplary counterions include monovalent inorganic cations, such as lithium, sodium, potassium, rubidium, cesium, silver, divalent inorganic cations, such as magnesium, calcium, nickel, zinc, iron copper, or manganese, and ammonium and alkylammonium counterions.
  • the counterion A + need not necessarily be the same throughout the molecule and comprise a combination of differing counterions, as readily understood by one of ordinary skill in the art.
  • the -OC(O)(alkyl) is chosen from -OC(O)(C 1 - C 21 alkyl), such as -OC(O)(CrCi 1 alkyl), -OC(O)(C 1 -C 5 alkyl), Or -OC(O)(C 1 - C 3 alkyl).
  • the -OC(O)(C 1 -C 2I alkyl) can be referred to as a C 2 -C 22 ester of a carboxyalkylcellulose ester.
  • the carboxyalkylcellulose ester is chosen from carboxymethylcellulose esters.
  • Exemplary carboxyalkylcellulose esters include, but are not limited to carboxymethylcellulose acetate butyrate (CMCAB) (such as CMCAB-641-0.5 from Eastman Chemical Company), high solids CMCAB (HS- CMCAB), carboxymethylcellulose butyrate (CMCB), carboxymethylcellulose acetate propionate (CMCAP), high solids CMCAP (HS-CMCAP), carboxymethylcellulose propionate (CMCP), carboxymethylcellulose acetate (CMCA), carboxymethylcellulose acetate isobutryate (CMCAiB), carboxymethylcellulose isobutryate (CMCiB), carboxymethylcellulose acetate butyrate succinate, carboxymethylcellulose acetate butyrate maleate, carboxymethylcellulose acetate butyrate trimellitate.
  • CMCAB carboxymethylcellulose acetate butyrate
  • CMCAP carboxymethylcellulose acetate propionate
  • CMCP carboxymethylcellulose
  • the at least one carboxyalkylcellulose ester is carboxymethylcellulose propionate having a degree of substitution per anhydroglucose of -OC(O)CH 2 CH 3 ranging from 1.5 to 2.7. In another embodiment, the at least one carboxyalkylcellulose ester is carboxymethylcellulose butyrate having a degree of substitution per anhydroglucose Of -OC(O)CH 2 CH 2 CH 3 ranging from 1.5 to 2.7.
  • the at least one carboxyalkylcellulose ester is carboxymethylcellulose acetate propionate having a degree of substitution per anhydroglucose of -OC(O)CH 3 ranging from 0.1 to 2.65 and a degree of substitution per anhydroglucose Of -OC(O)CH 2 CH 2 H 3 ranging from 0.1 to 2.6.
  • the at least one carboxyalkylcellulose ester is carboxymethylcellulose acetate butyrate having a degree of substitution per anhydroglucose Of -OC(O)CH 3 ranging from 0.1 to 1.65 and a degree of substitution per anhydroglucose Of -OC(O)CH 2 CH 2 H 3 ranging from 0.1 to 2.6.
  • the medium is chosen from water, acidic aqueous buffers, neutral aqueous buffers, basic aqueous buffers, and natural and simulated bodily fluids, such as gastric fluid (with or without pepsin), or intestinal fluid (with or without pancreatin).
  • the medium is chosen from pharmaceutically acceptable media.
  • “low solubility”, “poorly soluble”, and “poorly water soluble” are indicated by the Biopharmaceutics Classification System (BCS).
  • BCS Biopharmaceutics Classification System
  • the bioavailability of a drug may be influenced by at least two factors: solubility and permeability of a drug or agent.
  • the Biopharmaceutics Classification System may be used to distinguish between classes of drugs based on the solubility and permeability of the drugs in vivo.
  • the Biopharmaceutics Classification system provides four cases (or classes) of drugs. These cases (or classes) are defined as: Class 1 , high solubility-high permeability drugs; Class 2, low solubility-high permeability drugs; Class 3, high solubility-low permeability drugs; and Class 4, low solubility-low permeability drugs.
  • the at least one pharmaceutically active agent belongs to class 2, i.e., low solubility-high permeability drugs, according to the BCS.
  • the at least one pharmaceutically active agent belongs to class 4, i.e., low solubility-low permeability drugs, according to the BCS.
  • low solubility In another embodiment, “low solubility”, “poorly soluble”, and “poorly water soluble” are defined as one that requires at least 10,000 mL of water to dissolve 1 g of the agent.
  • the composition comprises a solid dispersion (also known as solid solution), i.e., the at least one pharmaceutically active agent is dispersed in a polymeric carrier.
  • a solid dispersion also known as solid solution
  • the polymeric carrier may disrupt the crystal structure of the drug, thereby reducing the crystal lattice energy. The energy required to dissolve the drug substance can be reduced, which may result in increased dissolution rates, and thus, the increased bioavailability of the pharmaceutically active agent.
  • the pharmaceutically active agent in the solid dispersion substantially all crystallinity of the pharmaceutically active agent is suppressed by the polymeric carrier.
  • the pharmaceutically active agent has a percent crystallinity of less than 20%, such as a percent crystallinity of less than 15%, less than 10%, less than 5%, less than 3%, or less than 1 %.
  • the agent is amorphous.
  • no crystallinity is detected by x-ray in the solid dispersion containing the pharmaceutically active agent and the polymeric carrier.
  • the polymeric carrier comprises the at least one carboxyalkylcellulose ester.
  • the carboxyalkylcellulose ester carrier can be blended with other conventional carriers, such as hydrophilic compounds or polymers.
  • exemplary carriers include physiologically inert compounds that are sometimes water soluble, e.g., polyethylene glycols, such as those disclosed in U.S. Patent No. 6,197,787.
  • cellulose and its derivatives such as microcrystalline cellulose (MCC), cellulose acetate butyrate (CAB), methylcellulose, polyethylene glycol, polypropylene glycol, copolymers of polyethylene glycol and polypropylene glycol, poly(vinylpyrrolidone), ethyl cellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, carboxymethyl cellulose, carboxymethylethyl cellulose, starch, dextran, dextrin, chitosan, collagen, gelatin, bromelain, cellulose acetate, unplasticized cellulose acetate, plasticized cellulose acetate, reinforced cellulose acetate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropylmethylcellulose, hydroxypropylmethyl-cellulose phthalate, hydroxypropylmethylcellulose acetate succinate, hydroxypropylmethylcellulose acetate trim
  • Solid dispersions can be prepared by any method known in the art, including co-evaporation (spray drying, rotovapping, film casting, etc.), freeze drying (lyophilizing), co-precipitation (flake precipitation, powder precipitation, etc), melt blending, melt extrusion, co-grinding and roll mixing, and solvent-free processes.
  • Two exemplary methods include the fusion technique and the solvent technique.
  • the drug is dissolved in a molten carrier (the carboxyalkylcellulose ester) and the mixture cooled to form a solid.
  • a molten carrier the carboxyalkylcellulose ester
  • the solvent technique drug and carrier are dissolved in a solvent, followed by removal of the solvent by evaporation, spray drying, freeze drying, or co- precipitation.
  • the preparation of a solid dispersion composition comprises weighing out a polymeric carrier, such as a carboxyalkylcellulose ester (e.g. CMCAB, CMCAP, or CMCA), into a suitable vessel and an appropriate solvent is added to the vessel to dissolve the carboxyalkylcellulose ester.
  • a polymeric carrier such as a carboxyalkylcellulose ester (e.g. CMCAB, CMCAP, or CMCA)
  • CMCAB carboxyalkylcellulose ester
  • CMCAP e.g. CMCAB, CMCAP, or CMCA
  • CMCA carboxyalkylcellulose ester
  • additives e.g. surfactants, dispersants, etc
  • All the components of the desired solid dispersion are combined into a single vessel and thoroughly mixed.
  • the solid dispersion is then generated by one of the following techniques: co-precipitation into a non-solvent (e.g.
  • co-precipitation is the general term used to describe the combination of a solution or mixture containing a polymeric carrier (e.g. a carboxyalkylcellulose ester) and a drug (e.g. a poorly soluble drug), and optionally one or more other additives dissolved in an organic solvent with an aqueous non-solvent to produce a precipitate that is an intimate mixture (i.e. solid dispersion) of the polymeric carrier, pharmaceutically active agent(s), and optionally one or more other additives from the organic solution/mixture.
  • a polymeric carrier e.g. a carboxyalkylcellulose ester
  • a drug e.g. a poorly soluble drug
  • Flake precipitation a process known to those skilled in the art of cellulose ester chemistry, can be accomplished by adding a thin stream of the polymer/drug/solvent mixture (i.e. dope) to the aqueous non-solvent. Then term flake precipitation comes from the typical appearance of the precipitate that is formed by the process.
  • process variables including temperature, rate of addition, mixing rate, concentration of solids in the organic mixture, pH of the nonsolvent, organic solvent content in the precipitate mixture, hardening time, etc) can be adjusted to modify the physical nature (i.e. morphology, particle size, etc.) of the co- precipitate, the composition of the co-precipitate, and likely the dissolution profile of the solid dispersion.
  • Powder precipitation a process known to those skilled in the art of cellulose ester chemistry, is accomplished by adding the aqueous non-solvent to the polymer/drug/solvent mixture (i.e. dope) with appropriate mixing and temperature.
  • the term powder precipitation comes from the typical appearance of the precipitate that is formed by the process.
  • process variables including temperature, rate of addition, mixing rate, concentration of solids in the organic mixture, pH of the nonsolvent, organic solvent content in the precipitate mixture, hardening time, etc) can be adjusted to modify the physical nature (i.e. morphology, particle size, etc.) of the co-evaporate, the composition of the co-evaporate, and likely the dissolution profile of the solid dispersion.
  • co-evaporation is the general term used to describe the removal of solvent from a solution or mixture containing a polymeric carrier (e.g. a carboxyalkylcellulose ester) and a drug (e.g. a poorly soluble drug), and optionally one or more other additives dissolved in a volatile organic solvent or mixture of solvents to produce a precipitate that is an intimate mixture (i.e. solid dispersion) of the non-volatile components of the organic solution/mixture.
  • a polymeric carrier e.g. a carboxyalkylcellulose ester
  • a drug e.g. a poorly soluble drug
  • Co-evaporation under reduced pressure can be accomplished by a number of processes including but not limited to rotary evaporation and vacuum distillation.
  • distillation at atmospheric pressure can be used to prepare solid dispersion compositions.
  • the solid dispersion compositions can be prepared by co-evaporation by film formation. Co-evaporation by film formation can be accomplished by casting a film of the drug/carrier/additive/solvent mixture and allowing film formation to occur upon evaporation of the solvent at room temperature and atmospheric pressure. Those skilled in the art recognize that there are numerous process ways to accomplish film formation from lab scale methods to commercial scale methods and that changing various process parameters such as rate of evaporation, temperature, pressure, and humidity can impact the morphology of the film that is formed and change the performance (i.e. the release profiles) of the solid dispersions prepared via this process. In one embodiment, the solid dispersion compositions can be prepared by spray drying. Those skilled in the art recognize that the selection of process parameters can be used to modify properties of the solid dispersions produced via this method.
  • compositions disclosed herein exhibit increased dissolution rates over that of the pharmaceutically active agent alone. In one embodiment, the compositions disclosed herein exhibit a more sustained release profile than that of the pharmaceutically active agent alone. In one embodiment, "sustained release” refers to a sustained delivery (i.e., substantially continuous release) of the pharmaceutically active agent over time, such as a time of at least 4 h, e.g., a time ranging from 4-24 h, from 12-24 h, from 6-12 h, or even greater than 24 h, e.g., 1-5 days.
  • compositions disclosed herein exhibit a near zero-order release profile wherein the pharmaceutically active agent alone releases almost immediately.
  • zero order release is a type of sustained release indicated by a substantially linear plot of released pharmaceutically active agent over time, where "substantially linear” refers to a correlation coefficient (R) of at least 0.8, for a given time, such as a correlation coefficient of at least 0.9, or at least 0.95.
  • the composition exhibits release of the pharmaceutically active agent at a target pH.
  • the target pH is at least 5, such as a pH of at least 6, or a pH of at least 6.5.
  • release of the pharmaceutically active agent is stopped or reduced to a very slow rate at gastric pH (e.g., approximately 1.2), whereas release as described herein occurs at intestinal pH (e.g., approximately 6.8).
  • the polymeric carrier is water-swellable, i.e., the polymeric carrier can expand in volume upon exposure to water, such as at pH levels approaching neutral or basic values.
  • the carboxy(Ci- C- 3 )alkylcellulose esters can be modified to obtain desired response to water and pH. For example, increasing the acid number of the carboxyalkylcellulose ester may produce a polymer that is more sensitive to water and ultimately could produce a water-soluble carboxy(Ci-C 3 )alkylcellulose ester.
  • a carboxyalkylcellulose ester may be made more water soluble by performing at least one of: increasing the hydroxyl content on the backbone, replacing longer chain esters with shorter chain esters (e.g. replace butyryl content with acetyl content), and/or reducing the molecular weight of the cellulose.
  • the at least one carboxyalkylcellulose ester has a low molecular weight, as described in WO 04/83253, the disclosure of which is incorporated herein by reference.
  • changing the composition of the carboxyalkylcellulose ester may affect the way it interacts with solvent, drugs, pharmaceutical additives and other polymers.
  • selection of the appropriate polymer composition for a specific drug and optional additives can be aided by the use of solubility parameters to determine the "compatibility" of the polymeric carrier, the drug, and the optional additives.
  • “Pharmaceutically active agent” refers to a biologically active organics, biological compounds, and combinations and blends thereof, that can treat or prevent a condition or disease.
  • the pharmaceutically active agent can be chosen from any suitable drug known in the art, such as those chosen from the classes of drugs including, for example, analgesics, anti-inflammatory agents, anthelmintics, anti-arrhythmic agents, antibiotics (including penicillins), anticoagulants, antidepressants, antidiabetic agents, antiepileptics, antihistamines, antihypertensive agents, antimuscarinic agents, antimycobacterial agents, antineoplastic agents, immunosuppressants, antithyroid agents, antiviral agents, anxiolytic sedatives (hypnotics and neuroleptics), astringents, beta-adrenoceptor blocking agents, blood products and substitutes, cardiac inotropic agents, contrast media, corticosteroids, cough suppressants (expectorants and mucolytics), diagnostic agents, diagnostic imaging agents, diuretics, dopaminergics (antiparkinsonian agents), haemostatics, immunological agents, lipid regulating agents, muscle relaxants,
  • analgesics and anti-inflammatory agents include, but are not limited to, aloxiprin, auranofin, azapropazone, benorylate, diclofenac, diflunisal, etodolac, fenbufen, fenoprofen calcim, flurbiprofen, ibuprofen, indomethacin, ketoprofen, meclofenamic acid, mefenamic acid, nabumetone, naproxen, oxyphenbutazone, phenylbutazone, piroxicam, sulindac.
  • anti-arrhythmic agents include amiodarone HCI, disopyramide, flecainide acetate, quinidine sulphate.
  • anti-bacterial and anti-pneumocystic agents include, but are not limited to, atovaquone, azithromycin, benethamine penicillin, cinoxacin, ciprofloxacin HCI, clarithromycin, clofazimine, cloxacillin, demeclocycline, doxycycline, erythromycin, ethionamide, imipenem, nalidixic acid, nitrofurantoin, rifampin, rifampicin, spiramycin, sulphabenzamide, sulphadoxine, sulphamerazine, sulphacetamide, sulphadiazine, sulphafurazole, sulfamethizole, sulphamethoxazole, sulphapyridine, tetracycline, trimethoprim.
  • Exemplary anti-coagulants include, but are not limited to, dicoumarol, dipyridamole, nicoumalone
  • anti-depressants include, but are not limited to, amoxapine, maprotiline HCI, mianserin HCL, nortriptyline HCI, trazodone HCL, trimipramine maleate.
  • anti-diabetics include, but are not limited to, acetohexamide, chlorpropamide, gliclazide, glipizide, glyburide, tolazamide, tolbutamide, troglitazone.
  • anti-epileptics include, but are not limited to, beclamide, carbamazepine, clonazepam, ethotoin, methoin, methsuximide, methylphenobarbitone, oxcarbazepine, paramethadione, phenacemide, phenobarbitone, phenytoin, phensuximide, primidone, sulthiame, valproic acid.
  • anti-fungal agents include, but are not limited to, amphotericin, butoconazole nitrate, clotrimazole, econazole nitrate, fluconazole, flucytosine, griseofulvin, itraconazole, ketoconazole, miconazole, natamycin, nystatin, posaconazole, sulconazole nitrate, terbinafine HCI, terconazole, tioconazole, undecenoic acid.
  • anti-gout agents include, but are not limited to, allopurinol, probenecid, sulphin-pyrazone.
  • anti-helmintics include, but are not limited to, albendazole, bephenium hydroxynaphthoate, cambendazole, dichlorophen, ivermectin, mebendazole, niclosamide, oxamniquine, oxfendazole, oxantel embonate, praziquantel, pyrantel embonate, thiabendazole.
  • anti-hypertensive agents include, but are not limited to, amlodipine, atenolol, benidipine, darodipine, dilitazem HCI, diazoxide, felodipine, guanabenz acetate, isradipine, minoxidil, nicardipine HCI, nifedipine, nimodipine, phenoxybenzamine HCI, prazosin HCI, reserpine, terazosin HCI, verapamil, verapamil HCI.
  • Exemplary anti-hypercholesterolemic, antihyperlipoproteinemic, and lipid regulating agents include, but are not limited to, atorvastatin, bezafibrate, clofibrate, etofibrate, fenofibrate, fluvastatin, gemfibrozil, lovastatin, pravastatin, probucol, simvastatin.
  • anti-malarials include, but are not limited to, amodiaquine, chloroquine, chlorproguanil HCI, halofantrine HCI, mefloquine HCI, proguanil HCI, pyrimethamine, quinine sulphate.
  • anti-migraine agents include, but are not limited to, dihydroergotamine mesylate, ergotamine tartrate, methysergide maleate, pizotifen maleate, sumatriptan succinate.
  • anti-muscarinic agents include, but are not limited to, atropine, benzhexol HCI, biperiden, ethopropazine HCI, hyoscyamine, mepenzolate bromide, oxyphencylcimine HCI, tropicamide.
  • anti-neoplastic agents and immunosuppressants include, but are not limited to.aminoglutethimide, amsacrine, azathioprine, busulphan, chlorambucil, cyclosporin, dacarbazine, docetaxel, estramustine, etoposide, irinotecan, lomustine, melphalan, mercaptopurine, methotrexate, mitomycin, mitotane, mitozantrone, paclitaxel, procarbazine HCI, rapamycin, tamoxifen, tamoxifen citrate, testolactone.
  • Exemplary anti-osteoporotic agents include, but are not limited to, raloxifene.
  • Exemplary anti-protazoal agents include, but are not limited to, benznidazole, clioquinol, decoquinate, diiodohydroxyquinoline, diloxanide furoate, dinitolmide, furzolidone, metronidazole, nimorazole, nitrofurazone, ornidazole, tinidazole.
  • Exemplary anti-thyroid agents include, but are not limited to, carbimazole, propylthiouracil.
  • anti-viral agents include, but are not limited to, acyclovir, nelfinavir, nevirapine, saquinavir.
  • Exemplary anxiolytic, sedatives, hypnotics and neuroleptics include, but are not limited to, alprazolam, amylobarbitone, barbitone, bentazepam, bromazepam, bromperidol, brotizolam, butobarbitone, carbromal, chlordiazepoxide, chlormethiazole, chlorpromazine, clobazam, clotiazepam, clozapine, diazepam, droperidol, ethinamate, flunanisone, flunitrazepam, fluopromazine, flupenthixol decanoate, fluphenazine decanoate, flurazepam, haloperidol, lorazepam, lormetazepam, medazepam, meprobamate, methaqualone, midazolam, nitrazepam, oxazepam, pentobarbit
  • Exemplary ⁇ -Blockers include, but are not limited to, acebutolol, alprenolol, atenolol, labetalol, metoprolol, nadolol, oxprenolol, pindolol, propranolol.
  • Exemplary cardiac inotropic agents include, but are not limited to, amrinone, digitoxin, digoxin, enoximone, lanatoside C, medigoxin.
  • corticosteroids include, but are not limited to, beclomethasone, betamethasone, betamethasone-17-valerate, budesonide, cortisone acetate, desoxymethasone, dexamethasone, fludrocortisone acetate, flunisolide, flucortolone, fluticasone propionate, hydrocortisone, hydrocortisone-21- hemisuccinate, methylprednisolone, prednisolone, prednisone, triamcinolone.
  • Exemplary diuretics include, but are not limited to, acetazolamide, amiloride, bendrofluazide, bumetanide, chlorothiazide, chlorthalidone, ethacrynic acid, frusemide, metolazone, spironolactone, triamterene.
  • anti-parkinsonian agents include, but are not limited to, bromocriptine mesylate, lysuride maleate.
  • Exemplary gastro-intestinal agents include, but are not limited to, bisacodyl, cimetidine, cisapride, diphenoxylate HCI, domperidone, famotidine, loperamide, mesalazine, nizatidine, omeprazole, ondansetron HCL, ranitidine HCI, sulphasalazine.
  • histamine H-Receptor antagonists include, but are not limited to, acrivastine, astemizole, cinnarizine, cyclizine, cyproheptadine HCI, dimenhydrinate, flunarizine HCI, loratadine, meclozine HCI, oxatomide, terrenadine.
  • Exemplary nitrates and other anti-anginal agents include, but are not limited to, amyl nitrate, glyceryl trinitrate, isosorbide dinitrate, isosorbide mononitrate, pentaerythritol tetranitrate.
  • Exemplary nutritional agents include, but are not limited to, betacarotene, vitamin A, vitamin B 2 , vitamin D, vitamin E, vitamin K.
  • opioid analgesics include, but are not limited to, codeine, dextropropyoxyphene, diamorphine, dihydrocodeine, meptazinol, methadone, morphine, nalbuphine, pentazocine.
  • Exemplary hormones include, but are not limited to, clomiphene citrate, danazol, ethinyloestradiol, medroxyprogesterone acetate, mestranol, methyltestosterone, norethisterone, norgestrel, oestradiol, conjugated oestrogens, progesterone, stanozolol, stiboestrol, testosterone, testosterone propionate, tibolone, thyroxine.
  • Exemplary stimulants include, but are not limited to, amphetamine, dexamphetamine, dexfenfluramine, fenfluramine, mazindol.
  • Exemplary diagnostics agents include, but are not limited to, iopanoic acid.
  • the pharmaceutically active agent is chosen from phenytoin, carbamazepine, glyburide, and griseofulvin.
  • the pharmaceutically active agent is chosen from those intended for oral administration.
  • a description of these classes of drugs and a listing of species within each class can be found in Martindale, the Extra Pharmacopoeia, Thirty-fourth Edition, the Pharmaceutical Press, London, 2005, the disclosure of which is incorporated herein by reference.
  • the drug substances are commercially available and/or can be prepared by techniques known in the art.
  • nutraceuticals and dietary supplements can also be included, such as those disclosed in, for example, Roberts et al., Nutraceuticals: The Complete Encyclopedia of Supplements, Herbs, Vitamins, and healing Foods (American Nutraceutical Association, 2001), which is specifically incorporated by reference.
  • a nutraceutical or dietary supplement, also known as phytochemicals or functional foods, is generally any one of a class of dietary supplements, vitamins, minerals, herbs, or healing foods that have medical or pharmaceutical effects on the body.
  • nutraceuticals or dietary supplements include, but are not limited to, folic acid, fatty acids (e.g., DHA and ARA), fruit and vegetable extracts, vitamin and mineral supplements, phosphatidylserine, lipoic acid, melatonin, glucosamine/chondroitin, Aloe Vera, Guggul, glutamine, amino acids (e.g., iso-leucine, leucine, lysine, methionine, phenylanine, threonine, tryptophan, and valine), green tea, lycopene, whole foods, food additives, herbs, phytonutrients, antioxidants, flavonoid constituents of fruits, evening primrose oil, flax seeds, fish and marine animal oils, and probiotics. Nutraceuticals and dietary supplements also include bio-engineered foods genetically engineered to have a desired property, also known as pharmafoods.
  • the pharmaceutical composition can include at least one pharmaceutically acceptable additive, binding agents, filling agents, lubricating agents, suspending agents, sweeteners, flavoring agents, preservatives, buffers, wetting agents, disintegrants, effervescent agents, surfactants, plasticizers, and other excipients.
  • excipients are known in the art.
  • Exemplary binding agents include but not exclusively, carbohydrates, starches in native or treated form, lipids, waxes and fats.
  • filling agents are lactose monohydrate, lactose anhydrous, mannitol, and various starches
  • binding agents are various celluloses and cross-linked polyvinylpyrrolidone, microcrystalline cellulose, such as Avicel ® PH 101 and Avicel ® PH 102, microcrystalline cellulose, and silicified microcrystalline cellulose (SMCC).
  • Suitable lubricants including agents that act on the flowability of the powder to be compressed, are colloidal silicon dioxide, such as Aerosil® 200; talc, stearic acid, magnesium stearate, calcium stearate, and silica gel.
  • sweeteners are any natural or artificial sweetener, such as sucrose, xylitol, sodium saccharin, cyclamate, aspartame, and acsulfame.
  • flavoring agents examples include Magnasweet® (trademark of MAFCO), bubble gum flavor, and fruit flavors, and the like.
  • Exemplary flavoring agents include, but are not limited to, Magnasweet® (trademark of MAFCO), bubble gum flavor, and fruit flavors.
  • preservatives are potassium sorbate, methylparaben, propylparaben, benzoic acid and its salts, other esters of parahydroxybenzoic acid such as butylparaben, alcohols such as ethyl or benzyl alcohol, phenolic compounds such as phenol, or quaternary compounds such as benzalkonium chloride.
  • Suitable diluents include pharmaceutically acceptable inert fillers, such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures of any of the foregoing.
  • diluents include microcrystalline cellulose, such as Avicel® PH 101 and Avicel® PH 102; lactose such as lactose monohydrate, lactose anhydrous, and Pharmatose® DCL21 ; dibasic calcium phosphate such as Emcompress®; mannitol; starch; sorbitol; sucrose; and glucose.
  • microcrystalline cellulose such as Avicel® PH 101 and Avicel® PH 102
  • lactose such as lactose monohydrate, lactose anhydrous, and Pharmatose® DCL21
  • dibasic calcium phosphate such as Emcompress®
  • mannitol starch
  • sorbitol sucrose
  • glucose glucose
  • Exemplary disintegrants include lightly crosslinked polyvinyl pyrrolidone, corn starch, potato starch, maize starch, and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate, and mixtures thereof.
  • Exemplary effervescent agents are effervescent couples such as an organic acid and a carbonate or bicarbonate. Suitable organic acids include, for example, citric, tartaric, malic, fumaric, adipic, succinic, and alginic acids and anhydrides and acid salts.
  • Suitable carbonates and bicarbonates include, for example, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate, magnesium carbonate, sodium glycine carbonate, L- lysine carbonate, and arginine carbonate.
  • plasticizers include plasticizers that can be used in this invention include diethyl phthalate, triacetin, triethyl citrate, PEG 400, castor oil, propylene glycol, glycerin, low-molecular weight polyethylene glycols, surfactants, and organic acid esters, actetyltributyl citrate, acetyltriethyl citrate, benzyl benzoate, chlorobutanol, diacetylated monoglycerides, dibutyl sebacate, mineral oil and lanolin alcohols, petrolatum and lanolin alcohols.
  • Additional examples of plasticizers include carbohydrate and polyol esters such as but not limited to those described in US Patent applications
  • glucose pentaacetate ⁇ -glucose pentaacetate, ⁇ -glucose pentapropionate, ⁇ -
  • glucose pentapropionate ⁇ -glucose pentabutyrate and ⁇ -glucose pentabutyrate
  • quaternary ammonium compounds that might be used in this invention include di- ⁇ / ⁇ alkyl(C 8 -C- ⁇ 8 from coconut oil) dimethyl ammonium chloride, dimethyl dialkyl ammonium chloride, and poly(divinylbenzene-co- tirmethyl(vinylbenzyl)ammoniur ⁇ ) chloride).
  • antioxidants such as tocopherol, tocopherol acetate, ascorbyl palmitate, ascorbic acid, butylhydroxytoluene, butylhydroxyanisole and propyl gallate
  • pH stabilizers such as citric acid, tartaric acid, fumaric acid, acetic acid, glycine, arginine, lysine and potassium hydrogen phosphate
  • thickeners/suspending agents such as hydrogenated vegetable oils, beeswax, colloidal silicon dioxide, gums, celluloses, silicates, bentonite
  • flavouring agents such as cherry, lemon and aniseed flavors
  • sweeteners such as aspartame, saccharin and cyclamates; etc.
  • the at least one additive is chosen from Vitamin E TPGS, sucrose acetate isobutyrate (SAIB), glucose pentapropionate (GPP), diethyl phthalate (DEP), triacetin, polyoxyethyenesorbitan monooleate (T ween 80) or sodium dodecylsulfate (SDS).
  • the at least one additive is chosen Vitamin E TPGS, SAIB, glucose pentapropionate, DEP, triacetin, Tween 80 or sodium dodecylsulfate, lactose monohydrate, lactose anhydrous, mannitol, and various starches;
  • binding agents are various celluloses and cross-linked polyvinylpyrrolidone, microcrystalline cellulose, such as Avicel® PH 101 and Avicel® PH102, microcrystalline cellulose, silicified microcrystalline cellulose (SMCC), colloidal silicon dioxide, such as Aerosil® 200; talc, stearic acid, magnesium stearate, calcium stearate, silica gel, sucrose, xylitol, sodium saccharin, cyclamate, aspartame, and acsulfame.
  • flavoring agents are Magnasweet® (trademark of MAFCO), bubble gum flavor, fruit flavors, potassium sorbate, methylparaben, propylparaben, benzoic acid and its salts, other esters of parahydroxybenzoic acid such as butylparaben, alcohols such as ethyl or benzyl alcohol, phenolic compounds such as phenol, or quaternary compounds such as benzalkonium chloride, microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures of any of the foregoing.
  • MAFCO methylparaben
  • propylparaben benzoic acid and its salts
  • other esters of parahydroxybenzoic acid such as butylparaben
  • alcohols such as ethyl or benzyl alcohol
  • phenolic compounds such as phenol
  • quaternary compounds such as benzalkonium chloride
  • microcrystalline cellulose lactose
  • lactose dibasic calcium
  • diluents include microcrystalline cellulose, such as Avicel® PH101 and Avicel® PH102, lactose such as lactose monohydrate, lactose anhydrous, and Pharmatose® DCL21 , dibasic calcium phosphate such as Emcompress®, mannitol, starch, sorbitol, sucrose, glucose, lightly crosslinked polyvinyl pyrrolidone, corn starch, potato starch, maize starch, and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate,
  • Suitable organic acids include, for example, citric, tartaric, malic, fumaric, adipic, succinic, and alginic acids and anhydrides and acid salts.
  • Suitable carbonates and bicarbonates include, for example, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate, magnesium carbonate, sodium
  • cyclodextrin and ⁇ -cyclodextrin. More specifically examples of cyclodextrin
  • derivatives include hydroxypropyl- ⁇ -cyclodextrin, hydroxypropyl- ⁇ -cyclodextrin,
  • petrolatum and lanolin alcohols petrolatum and lanolin alcohols, glucose pentapropionate, ⁇ -glucose
  • amines and amino derivatives amine- containing polymers, chitosan, tocopherol, tocopherol acetate, ascorbyl palmitate, ascorbic acid, butylhydroxytoluene, butylhydroxyanisole and propyl gallate; pH stabilizers such as citric acid, tartaric acid, fumaric acid, acetic acid, glycine, arginine, lysine
  • the pharmaceutical composition can take a variety of forms, including, for example, those chosen from tablets, caplets, hard and soft gelatin capsules, non-gelatin-based capsules, powders, and sprinkles.
  • the composition can be formulated into an oral dosage form.
  • the composition can be formulated for rectal, intravaginal, injectable, pulmonary, nasal, buccal, topical, local, intracisternal, intraperitoneal, ocular, aural, buccal spray, or nasal spray administration.
  • the composition when the pharmaceutical composition is in the form of a tablet, the composition is sufficiently compressible for tablet formation.
  • the composition can sustain a compression force of at least 10 psi for at least 10 seconds, such as a compression force of at least 100 psi for at least 10 seconds, such as a compression force of at least 1000 psi for at least 10 seconds.
  • the formulations disclosed herein can be made using at least one method chosen from spray drying, spray granulation, fluid bed granulation, high shear granulation, fluid bed drying, lyophilization, tableting, jet milling, pin milling, wet milling, rotogranulation, and spray coating.
  • the composition comprises:
  • the at least one pharmaceutically active agent in an amount ranging from 0.1 to 99 weight percent, based on the total weight (a) and (b) in said composition;
  • an organic solvent including but not limited to acetone, ethanol, ethyl acetate, dichloromethane, dimethyl sulfoxide, or water, or a solvent mixture; wherein the total weight of (a) and (b) is about 5 to 95 weight percent of the total weight of (a), (b), (c), and (d).
  • the composition comprises: (a) about 0.1 to about 99 weight percent, based on the total weight (a) and (b) in said composition, of at least one carboxy(C-i - C 3 )alkylcellulose ester as disclosed herein having an inherent viscosity of about 0.20 to 0.70 dL/g, as measured in a 60/40 (wt./wt.) solution of phenol/tetra-chloroethane at 25 °C, a degree of substitution per anhydroglucose unit of carboxy ⁇ - C 3 )alkyl of greater than 0.2 to about 0.75, and a degree of substitution per anhydroglucose unit of C 2 - C 2O esters of about 1.5 to about 2.70;
  • (c) about 0 to about 50 weight percent, based on the total weight of (a), (b), and (c) in said composition, of at least one additive selected from plasticizers, flow aids, binding agents, filling agents, lubricating agents, suspending agents, sweeteners, flavoring agents, preservatives, buffers, wetting agents, disintegrants, effervescent agents, etc.
  • aqueous and/or organic solvents including but not limited to acetone, ethanol, ethyl acetate, dichloromethane, dimethyl sulfoxide, or water, or a solvent mixture
  • the at least one carboxy(Ci - C 3 )alkylcellulose ester is chosen from a C 2 -C 4 ester of a carboxy(Ci - C 3 )alkylcellulose ester.
  • ingredient (a) in the compositions disclosed herein can comprise about 0.1 to about 99 weight percent, based on the total weight (a) and (b) in said composition, of a carboxymethylcellulose acetate butyrate, an inherent viscosity of about 0.20 to 0.70 dL/g, as measured in a 60/40 (wt./wt.) solution of phenol/tetra-chloroethane at 25 °C, a degree of substitution per anhydroglucose unit of carboxy(Ci - C 3 )alkyl of greater than 0.2 to about 0.75, and a degree of substitution per anhydroglucose unit of butyrate esters of about 1.5 to about 2.70, and a degree of substitution per anhydroglucose unit of acetate esters of about 0.1 to about 2.0, and a degree of substitution of hydroxyl groups of from about 0.01 to about 1.5.
  • ingredient (a) in the compositions disclosed herein can comprise about 0.1 to about 99 weight percent, based on the total weight (a) and (b) in said composition, of a carboxymethylcellulose acetate propionate, an inherent viscosity of about 0.20 to 0.70 dL/g, as measured in a 60/40 (wt./wt.) solution of phenol/tetra-chloroethane at 25 °C, a degree of substitution per anhydroglucose unit of carboxy(Ci - C 3 )alkyl of greater than 0.2 to about 0.75, and a degree of substitution per anhydroglucose unit of propionate esters of about 1.5 to about 2.70, and a degree of substitution per anhydroglucose unit of acetate esters of about 0.1 to about 2.0, and a degree of substitution of hydroxyl groups of from about 0.01 to about 1.5.
  • ingredient (a) in the compositions disclosed herein can comprise (a) about 0.1 to about 99 weight percent, based on the total weight (a) and (b) in said composition, of a carboxymethylcellulose acetate, an inherent viscosity of about 0.20 to 0.70 dL/g, as measured in a 60/40 (wt./wt.) solution of phenol/tetra-chloroethane at 25 0 C, a degree of substitution per anhydroglucose unit of carboxymethyl groups of greater than 0.2 to about 0.75, and a degree of substitution per anhydroglucose unit of acetate esters of about 1.5 to about 2.70, and a degree of substitution of hydroxyl groups of from about 0.01 to about 1.5.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate butyrate (CMCAB), a BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate propionate (CMCAP) and at least one BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate (CMCA) and at least one BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate isobutyrate (CMCAiB), and at least one BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising a C 2 -C20 alkyl acid (CMC C 2 -C 20 Ester), and at least one BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising at least one carboxymethylcellulose mixed ester of at least one C 2 -C 2O alkyl acid (CMC C 2 - C 2 o mixed ester) and a BCS Class 2 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate butyrate (CMCAB), a BCS Class 4 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate propionate (CMCAP) and at least one BCS Class 4 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate (CMCA) and at least one BCS Class 4 drug.
  • the composition comprises a solid dispersion comprising carboxymethylcellulose acetate isobutyrate (CMCAiB), and at least one BCS Class 4 drug.
  • the composition comprises a solid dispersion comprising a C2-C2 0 alkyl acid (CMC C 2 -C 20 Ester), and at least one BCS Class 4 drug.
  • the composition comprises a solid dispersion comprising at least one carboxymethylcellulose mixed ester of at least one C2-C20 alkyl acid (CMC C 2 - C 2O mixed ester) and a BCS Class 4 drug.
  • Another embodiment disclosed herein provides a method of treating a mammal in need thereof with a pharmaceutical composition, comprising: administering to the mammal in need of treatment the pharmaceutical composition comprising: a therapeutically effective amount of at least one pharmaceutically active agent having low solubility in a medium, and at least one carboxyalkylcellulose ester and pharmaceutically acceptable salts thereof comprising an anhydroglucose repeat unit having the structure:
  • R 1 -R 6 are each independently selected from -OH, -OC(O)(alkyl), and -0(CHb) x C(O)OH , and pharmaceutically acceptable salts thereof, wherein x ranges from 1-3,
  • - a degree of substitution per anhydroglucose of - OH ranges from 0.1 to 0.7
  • - a degree of substitution per anhydroglucose of -OC(O)(alkyl) ranges from 0.1 to 2.7
  • treatment and its cognates (e.g.,
  • therapeutic method refers to both therapeutic treatment and prophylactic/preventative measures. Those in need of treatment may include humans or animals already having a particular medical disease as well as those at risk for the disease (i.e., those who are likely to ultimately acquire the disorder). A therapeutic method results in the prevention or amelioration of symptoms or an otherwise desired biological outcome and may be evaluated by improved clinical signs, delayed onset of disease, reduced/elevated levels of lymphocytes and/or antibodies, etc.
  • compositions described herein may be varied so as to obtain an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular patient, compositions, and mode of administration.
  • therapeutically effective dose and “therapeutically effective amount” refer to that amount of a compound that results in prevention or amelioration of symptoms in a patient or a desired biological outcome, e.g., improved clinical signs, delayed onset of disease, reduced/elevated levels of lymphocytes and/or antibodies, etc.
  • the effective amount can be determined as described herein.
  • the selected dosage level will depend upon the activity of the particular compound, the route of administration, the severity of the condition being treated, and the condition and prior medical history of the patient being treated.
  • the data obtained from the assays can be used in formulating a range of dosage for use in humans.
  • dosage levels of about 0.1 ⁇ g/kg to about 50 mg/kg can be administered topically, orally or intravenously to a mammalian patient.
  • Other dosage levels range from about 1 ⁇ g/kg to about 20 mg/kg, from about 1 ⁇ g/kg to about 10 mg/kg, from about 1 ⁇ g/kg to about 1 mg/kg, from 10 ⁇ g/kg to 1 mg/kg, from 10 ⁇ g/kg to 100 ⁇ g/kg, from 100 ⁇ g to 1 mg/kg, and from about 500 ⁇ g/kg to about 5 mg/kg per day.
  • the effective daily dose may be divided into multiple doses for purposes of administration, e.g., two to four separate doses per day.
  • the pharmaceutical composition can be administered once per day.
  • Vitamin E TPGS, NF Grade Eastman Chemical Company, Batch #'s 30035000 and 40008000
  • SAIB Sucrose acetate isobutyrate
  • Teween 80 Polyoxyethylenesorbitan monooleate
  • Griseofulvin (Sigma-Aldrich, Cat. # 64753-25G, Batch # 083K1219) Azithromycin (LKT Laboratories, Cat. # A9834, Lot # 2393101 )
  • Glyburide RS (USP, Cat. # 1295505, Lot # G1C347)
  • Azithromycin RS (USP, Cat. # 1046056, Lot # H0C212)
  • VCaps Capsugel, size OCS, Lot #630311
  • a GC method is used to determine acetyl, propionyl, and butyryl, rather than NMR, because the methylene of the carboxyl(Ci-C 3 )alkyl group could not be easily separated from the ring protons of the cellulose backbone.
  • the DS values were calculated by converting the acid number to percent carboxymethyl and using this along with the GC weight percents of acetyl, propionyl, and butyryl.
  • the acetyl, propionyl, and butyryl weight percents were determined by a hydrolysis GC method.
  • a hydrolysis GC method In this method, about 1 g of ester was weighed into a weighing bottle and dried in a vacuum oven at 105 °C. for at least 30 minutes. Then 0.500 ⁇ 0.001 g of sample was weighed into a 250 mL Erlenmeyer flask. To this flask was added 50 mL of a solution of 9.16 g isovaleric acid, 99%, in 2000 mL pyridine. This mixture was heated to reflux for about 10 minutes, after which 30 mL of isopropanolic potassium hydroxide solution is added. This mixture was heated at reflux for about 10 minutes.
  • Ci concentration of I (acyl group)
  • Ci ((Fi*Ai)/Fs*As))*R*100
  • the GC method was used, along with NMR, to determine weight % acetyl, propionyl, and butyryl, and the method used is indicated.
  • the acid number of the carboxy(C 1 -C 3 )alkylcellulose esters was determined by titration as follows. An accurately weighed aliquot (0.5-1.0 g) of the carboxy(Ci-C 3 )alkylcellulose ester was mixed with 50 ml_ of pyridine and stirred. To this mixture was added 40 ml_ of acetone followed by stirring. Finally, 20 ml_ of water was added and the mixture stirred again. This mixture was titrated with 0.1 N sodium hydroxide in water using a glass/combination electrode. A blank containing 50 ml_ of pyridine, 40 mL of acetone, and 20 ml_ of water was also titrated. The acid number was calculated as follows where:
  • Ep mL NaOH solution to reach end point of sample
  • IV The inherent viscosity (IV) of the cellulose esters and carboxy(C-i- C 3 )alkylcellulose esters described herein, except where indicated otherwise, was determined by measuring the flow time of a solution of known polymer concentration and the flow time of a solvent-blank in a capillary viscometer, and then calculating the IV. IV is defined by the following equation:
  • (n) lnherent Viscosity at 25 0 C. at a polymer concentration of 0.50 g/l 00 ml_ of solvent.
  • Samples were prepared to a concentration of 0.50 g per 100 mL of solvent (60 % phenol and 40 % 1 ,1 ,2,2-tetrachloroethane, or "PM95,” by weight).
  • the sample (0.25 g) was weighed into a culture tube containing a stir bar. 50.0 mL of 60 % phenol and 40 % 1 ,1 ,2,2-tetrachloroethane by weight (also described in the application as "PM95") is added.
  • the mixture was placed in a heater and heated with stirring (300 rpm) to 125 °C. (7 minutes to reach the target temperature and 15 minute hold at 125 0 C).
  • the sample was allowed to cool to room temperature (25 °C.) and was then filtered and placed in the viscometer (Model AVS 500-Schott America, Glass & Scientific Products, Inc., Yonkers, N.Y.). IV was calculated according to the equation above.
  • THF The molecular weight distributions of cellulose ester samples indicated as being tested by GPC with THF as a solvent were determined at ambient temperature in Burdick and Jackson GPC-grade THF stabilized with BHT, at a flow rate of 1 mL/min. All other samples were determined using GPC with NMP as a solvent, as set forth in Method 2 below. Sample solutions were prepared by dissolution of about 50 mg of polymer in 10 ml_ of THF, to which 10 ⁇ l_ of toluene was added as a flow-rate marker.
  • NMP The molecular weight distributions of all samples not otherwise indicated were determined by GPC with NMP as a solvent, as follows. The molecular weight distributions of cellulose ester samples were determined by gel permeation chromatography at 40 0 C. in Burdick and Jackson N- Methylpyrrolidone with 1 % Baker glacial acetic acid by weight, at a flow rate of 0.8 mL/min. Sample solutions were prepared by dissolution of about 25 mg of polymer in 10 ml_ of NMP, to which 10 ⁇ L of toluene was added as a flow-rate marker. An autosampler was used to inject 20 ⁇ L of each solution onto a
  • UV 254 nm UV 254 nm (UV 214 nm can also be used for phenytoin or carbamazepine, UV 291 nm can also be used for griseofulvin); typically five signals were selected from UV 214 nm, 222 nm, 254 nm, 287 nm, 291 nm, and/or 325 nm were collected for each sample.
  • Retention times were typically between 1.5 and 2.5 minutes.
  • Detection Five signals selected from UV 210 nm, 214 nm, 220 nm, 230 nm, and 240 nm are collected for each sample. Determination of % Crystallinity by X-ray.
  • R wc/ws*ls/lc where we id the weight fraction of corundum, ws is the weight fraction of the species of interest, Ic is the net intensity of the major diffraction line of corrundum and is the net intensity of the major diffraction line of the drug or in the case of the polymers, the net intensity of the maximum of the amorphous scattering curve.
  • Samples were pelletized with a hydraulic press and the XRD pattern of the pellet was measured from 5 to 45 degree scattering angle. The net intensity of the maximum of the amorphous scatter from the polymer, Ip, and the net intensity of the major diffraction line of the drug, Id, were determined from the resulting scattering curve.
  • % crystalline drug (ld/Rd)/(ld/Rd+lp/Rp)x100 where Rd is the response factor for the drug and Rp is the response factor for the polymer.
  • Glyburide reference standard from USP was dried as directed (106 0 C for six hours), then approximately 20-25 mg of glyburide are added to a 25-mL volumetric flask and dissolved in DMSO or 55 % acetonitrile/ 45 % ammonium acetate, pH 5.25. The volume was diluted to 25 ml_. A set of standard dilutions were prepared either using 10-mL volumetric flasks or Rainin automatic pipetmen. Preparation of Dissolution Media
  • Simulated Intestinal Fluid, without pancreatin, pH 6.8 (SIF sp , pH 6.8) - Added monobasic potassium phosphate (KHaPO 4 , 34 g) to a 4000-mL beaker. Added deionized/polished water (2000 ml_) and mixed using a magnetic stir bar until the Kh ⁇ PO 4 is completely dissolved. Added 0.2 N sodium hydroxide (NaOH, 590 ml_) and stirred. The pH was adjusted to pH 6.8 ⁇ 0.1 using 0.2 N NaOH. The sample was diluted with deionized/polished water to a final volume of 5000 mL.
  • the SIFsp media was heated to ⁇ 45 °C in four 2000-mL Kimax bottles in an oven.
  • the sample was degassed according to USP protocol by filtering through a 0.45 ⁇ m membrane filter (Pall, Supor-450, 0.45 ⁇ m, 90 mm, part # 60200, Lot # 43214) and stirring under vacuum for 5 minutes.
  • the SGFsp media was heated to ⁇ 45 °C in four 2000-mL Kimax bottles in an oven.
  • the sample was degassed according to USP protocol by filtering through a 0.45 ⁇ m membrane filter (Pall, Supor-450, 0.45 ⁇ m, 90 mm, part # 60200, Lot # 43214) and stirring under vacuum for 5 minutes.
  • Dissolution Conditions #1 Dissolution studies were performed on a
  • Dissolution studies were performed on a Varian VK7025 Dissolution Apparatus equipped with a Varian VK8000 Fraction Collector using the following parameters: stir rate (75 rpm), sample size (5 imL), sample times (15 min, 30 min, 45 min, 1 hr, 1 hr 30 min, 2 hr, 2 hr 30 min, 3 hr, 3 hr 30 min, 4 hr), bath temperature (37.3 0 C), vessel temperature (37 °C), pump prime (60 sec), pump purge (60 sec), filter tips (10 ⁇ m).
  • the Buchi Model B-290/B295 Mini Spray Dryer is a lab-scale glass spray dryer with the capability to process flammable solvents.
  • a closed-loop solvent recovery system with online oxygen monitoring allows safe processing of flammable solvents.
  • Atomization is accomplished by a two-fluid nozzle.
  • the feed material was supplied to the nozzle by a built-in peristaltic pump.
  • the drying gas flow was co-current to atomization of the feed.
  • Product is isolated from the gas stream by a cyclone separator.
  • a bag filter was downstream of the cyclone to remove residual material from the exhaust gas stream.
  • the nitrogen supply valve was opened to provide an inert atmosphere to the dryer and the fan was switched on.
  • the system was inspected to determine if there were any leaks in the glassware that would allow air into the system. Once the oxygen level was below 5%, the inlet temperature was set and the heater was switched on.
  • the condenser temperature was set to the necessary temperature to allow removal of the solvent from the gas stream without freezing.
  • the atomization gas was set to the desired flow by adjusting the flow meter.
  • the feed material was inspected to ensure that the viscosity was suitable for adequate atomization and also to determine the need for filtration to remove insoluble materials. Once the desired inlet temperature was reached, the pump tubing was placed in the feed material and pump was switched on.
  • the pump speed was set low (--10%) and slowly increased if no problems were encountered.
  • clean solvent was pumped through the nozzle to prevent plugging.
  • the heater was switched off and the fan was allowed to run to cool the unit. Once the unit was cool, the fan and atomization gas was switched off.
  • the product collection container was removed and the product was transferred to a container. To maximize yield, the glassware was cleaned with a spatula and the product was collected and combined with that from the product container. Typical process conditions are given below.
  • Example 1 This Example describes the preparation of solid dispersions by co- precipitation (flake method).
  • Co-precipitation is the general term used to describe the combination of a solution or mixture containing a polymeric carrier (e.g. a carboxyalkylcellulose ester) and a pharmaceutically active agent, and optionally one or more other additives dissolved in an organic solvent with an aqueous non-solvent to produce a precipitate that is an intimate mixture (i.e. solid dispersion) of the non-volatile components of the organic solution/mixture.
  • the two co-precipitation methods used for the preparation of the compositions of this invention are flake precipitation and powder precipitation.
  • Flake precipitation a process known to those skilled in the art of cellulose ester chemistry, is accomplished by adding a thin stream of the polymer/drug/solvent mixture (i.e. dope) to the aqueous non-solvent.
  • the term flake precipitation comes from the typical appearance of the precipitate that is formed by the process.
  • process variables including but not limited to temperature, rate of addition, mixing rate, concentration of solids in the organic mixture, pH of the nonsolvent, organic solvent content in the precipitate mixture, hardening time, etc) can be adjusted to modify the physical nature (i.e. morphology, particle size, etc.) of the co-precipitate, the composition of the co-precipitate, and likely the dissolution profile of the solid dispersion.
  • an appropriate organic solvent or mixture of solvents e.g. acetone, methylene chloride, ethanol, etc.
  • a vessel typically a glass bottle
  • the desired amount of the polymer carrier in other examples, CMCAB, HPMCAS, PVP, or PEG
  • the solids content of the mixture is adjusted by addition of solvent of solvent blend to produce a mixture with the desired viscosity.
  • the drug substance is dissolved in an appropriate solvent or mixture of solvents (e.g.
  • one or more additives can be added to a third vessel and dissolved in an appropriate solvent.
  • the polymer solution, the drug solution, and if included the additive solution are combined and thoroughly mixed.
  • the polymer and drug solids and optional additives, if desired or required can be combined in a single vessel and then dissolved at the same time by the addition of an organic solvent or solvent mixture (This strategy is not always appropriate and should be tested on a case by case basis).
  • Co-precipitation is induced by pouring a small stream of the polymer/drug/additive solution into an excess of water, aqueous base, aqueous acid, or aqueous buffer solution with rapid mixing.
  • a ratio of 1 :3 organic to aqueous solution is appropriate to induce flake precipitation, but a larger excess of aqueous solution is often appropriate depending on the percent solids in the system and the nature of the organic solvent in use.
  • samples are stored in a desiccator or vacuum desiccator until needed.
  • This Example describes the preparation of solid dispersions by co- precipitation (powder method).
  • Powder precipitation a process known to those skilled in the art of cellulose ester chemistry, is accomplished by adding the aqueous non-solvent to the polymer/drug/solvent mixture (i.e. dope) with appropriate mixing and temperature. Then term powder precipitation comes from the typical appearance of the precipitate that is formed by the process.
  • process variables including but not limited to temperature, rate of addition, mixing rate, concentration of solids in the organic mixture, pH of the nonsolvent, organic solvent content in the precipitate mixture, hardening time, etc) can be adjusted to modify the physical nature (i.e. morphology, particle size, etc.) of the co- precipitate, the composition of the co-precipitate, and likely the dissolution profile of the solid dispersion..
  • an appropriate organic solvent or mixture of solvents e.g. acetone, methylene chloride, ethanol, etc.
  • a vessel typically a glass bottle
  • the desired amount of the polymer carrier in other examples, CMCAB, HPMCAS, PVP, or PEG
  • the solids content is adjusted by addition of solvent of solvent blend to produce a mixture with the desired viscosity.
  • the drug substance is dissolved in an appropriate solvent or mixture of solvents (e.g. acetone, methylene chloride, ethanol, dimethyl sulfoxide, etc.) in a separate vessel.
  • additional additive or additives are added to a third vessel and dissolved in an appropriate solvent.
  • the polymer solution, the drug solution, and if included the additive solution are combined and thoroughly mixed.
  • the polymer and drug solids and optional additives, if desired or required can be combined in a single vessel and then dissolved at the same time by the addition of an organic solvent or solvent mixture.
  • Co-precipitation is induced by slowly adding water, an aqueous base, an aqueous acid, or aqueous buffer solution to the polymer/drug/additive organic solution with rapid mixing.
  • a ratio of 1 :3 organic to aqueous solution is appropriate to induce powder precipitation, but a larger excess of aqueous solution is often appropriate depending on the percent solids in the system and the nature of the organic solvent in use.
  • the sample if filtered on a coarse fritted funnel, dried overnight at 45 0 C in a vacuum oven, and pulverized to a particle size of approximately 20 ⁇ m (typically less than 200 ⁇ m) in a cryogenic grinder.
  • the samples are stored in a desiccator or vacuum desiccator until needed.
  • This Example describes the preparation of solid dispersions by co- evaporation (reduced pressure method).
  • Co-evaporation is the general term used to describe the removal of solvent from a solution or mixture containing a polymeric carrier (e.g. a carboxyalkylcellulose ester) and a drug as disclosed herein, and optionally one or more other additives dissolved in a volatile organic solvent or mixture of solvents to produce a precipitate that is an intimate mixture (i.e. solid dispersion) of the non-volatile components of the organic solution/mixture.
  • the three co-evaporation methods used for the preparation of the compositions disclosed herein are rotary evaporation under reduced pressure, film formation (i.e. evaporation without mixing at atmospheric pressure), and spray drying.
  • an appropriate organic solvent or mixture of solvents e.g. acetone, methylene chloride, ethanol, etc.
  • a vessel typically a glass bottle
  • the desired amount of the polymer carrier in other examples, CMCAB, HPMCAS, PVP, or PEG
  • the solids content is adjusted by addition of solvent of solvent blend to produce a mixture with the desired viscosity.
  • the drug substance is dissolved in an appropriate solvent or mixture of solvents (e.g. acetone, methylene chloride, ethanol, dimethyl sulfoxide, etc.) in a separate vessel.
  • one or more additives are added to a third vessel and dissolved in an appropriate solvent.
  • the polymer solution, the drug solution, and if included the additive solution are combined and thoroughly mixed.
  • the polymer and drug solids and optional additives, if desired or required can be combined in a single vessel and then dissolved at the same time by the addition of an organic solvent or solvent mixture.
  • Co-evaporation is induced by removing the solvent from the system using a rotary evaporator, typically at 50 °C.
  • a rotary evaporator typically at 50 °C.
  • the sample is placed on a high vacuum line over night to remove as much residual solvent as possible.
  • the sample is removed from the round-bottomed flask by with a spatula.
  • the sample is then dried overnight at 45 °C in a vacuum oven and then pulverized to a particle size of approximately 20 ⁇ m in a cryogenic grinder.
  • the samples are stored in desiccator or vacuum desiccator until needed.
  • This Example describes the preparation of solid dispersions by co- evaporation via the film formation method.
  • An appropriate organic solvent or mixture of solvents e.g. acetone, methylene chloride, ethanol, etc.
  • a vessel typically a glass bottle
  • the desired amount of the polymer carrier in other examples, CMCAB, HPMCAS, PVP, or PEG
  • the solids content was adjusted by addition of solvent of solvent blend to produce a mixture with the desired viscosity.
  • the drug is dissolved in an appropriate solvent or mixture of solvents (e.g.
  • acetone, methylene chloride, ethanol, dimethyl sulfoxide, etc. in a separate vessel.
  • one or more additives are added to a third vessel and dissolved in an appropriate solvent.
  • the polymer solution, the drug solution, and if included the additive solution are combined and thoroughly mixed.
  • the polymer and drug solids and optional additives, if desired or required can be combined in a single vessel and then dissolved at the same time by the addition of an organic solvent or solvent mixture (This strategy is not always appropriate and should be tested on a case by case basis).
  • Co-evaporation is induced by removing the solvent from the system by pouring the sample into an appropriate vessel or onto a glass or metal sheet and allowing the solvent to slowly evaporate and a film to be formed.
  • a number of parameters can be controlled to influence the properties of the film formed.
  • the samples are poured into an evaporation dish and allowed to stand overnight while covered with a paper towel.
  • the sample is removed from the dish with a spatula.
  • the sample is then dried overnight at 45 "C in a vacuum oven and then pulverized to a particle size of approximately 20 ⁇ m (typically less than 200 ⁇ m) in a cryogenic grinder.
  • the samples are stored in a desiccator or vacuum desiccator until needed.
  • Examples 60-73 These Examples describe the preparation of ibuprofen/CMCAB solid dispersions and ibuprofen/CM CAB/PEG solid dispersions.
  • Solid dispersions of containing ibuprofen (IB), carboxymethylcellulose acetate butyrate (CMCAB), and optionally polyethylene glycol (PEG) were prepared by co-precipitation, the flake method of Example 1. Specific details of the preparation are listed in Table 5 below.
  • Solid dispersions or physical blends of phenytoin (Phe), a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB), hydroxyproplymethylcellulose acetate succinate (HPMCAS), or cellulose acetate phthalate (C-A-P)) and optionally an additive (Pz) (vitamin E TPGS (TPGS) or sucrose acetate isobutyrate (SAIB)) were prepared by the co- precipitation, flake method of Example (solid dispersions) or by physical mixing (physical blends). Specific details of the preparation are listed in Table 6 below.
  • phenytoin/polymer solid dispersions by spray drying as described in the Materials and Methods section.
  • Solid dispersions of phenytoin (Phe) and a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB), hydroxyproplymethylcellulose acetate succinate (HPMCAS), or cellulose acetate phthalate (C-A-P)) were prepared by spray drying. Specific details of the preparation are listed in Table 7, below.
  • Solid dispersions or physical blends of carbamazepine (Cbz), a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB)) and optionally an additive (Pz) (vitamin E TPGS (TPGS) or sucrose acetate isobutyrate (SAIB)) were prepared by co-precipitation, co-evaporation, lyophilization, or spray drying (see Examples 1-4 for details) (solid dispersions) or physical mixing (physical blends), as described in Table 8.
  • Solid dispersions or physical blends of nitrofurantoin (Nit), a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB)), and optionally an additive (Pz) (vitamin E TPGS (TPGS) or sucrose acetate isobutyrate (SAIB)) were prepared by co-precipitation, co-evaporation, lyophilization, or spray drying (see Examples 1-4 for details) (solid dispersions) or physical mixing (physical blends), as described in Table 9.
  • Solid dispersions or physical blends of glyburide (GIy), a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB)), and optionally an additive (Pz) (vitamin E TPGS (TPGS) or sucrose acetate isobutyrate (SAIB)) were prepared by co-precipitation, co-evaporation, lyophilization, or spray drying (see Examples 1-4 for details) (solid dispersions) or physical mixing (physical blends), as described in Table 10.
  • Glyburide was not soluble in acetone and thus DMSO was used to dissolve glyburide.
  • the glyburide/DMSO solution was added to the polymer/additive (optional) solution in acetone prior to formation of the solid dispersion.
  • Example 118 This Example describes the preparation of a glyburide/CMCAB solid dispersion by spray drying.
  • a solid dispersion of glyburide (GIy) and a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB)) was prepared by spray drying as described in Table 11 using the spray drying conditions described in the Materials and Methods section.
  • Glyburide was not soluble in acetone and thus DMSO was used to dissolve glyburide.
  • the glyburide/DMSO solution was added to the polymer/additive (optional) solution in acetone prior to formation of the solid dispersion. Examples 119-124
  • Solid dispersions of glyburide (GIy) and a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB) or hydroxypropylmethylcellulose acetate butyrate (HPMCAS)) were prepared by co- precipitation, flake method (see Example 1 for details) as described in Table 12.
  • CMCAB carboxymethylcellulose acetate butyrate
  • HPMCAS hydroxypropylmethylcellulose acetate butyrate
  • Solid dispersions of griseofulvin (Gris) and a polymer carrier (carboxymethylcellulose acetate butyrate (CMCAB), hydroxypropylmethylcellulose acetate butyrate (HPMCAS), or polyvinylpyrrolidone (PVP)) were prepared by co-precipitation, flake method, or co-evaporation (see Examples 1-4 for details) as described in Table 13.
  • CMCAB carboxymethylcellulose acetate butyrate
  • HPMCAS hydroxypropylmethylcellulose acetate butyrate
  • PVP polyvinylpyrrolidone
  • Gris griseofulvin
  • CMCAB carboxymethylcellulose acetate butyrate
  • SDS sodium dodecylsulfate
  • a Varian VK7025 dissolution apparatus and a Varian VK8000 autosampler were used for the dissolution studies using the following parameters: stir rate (50 rpm), sample size (5 mL), sample times (15 min, 30 min, 1 hr, 1hr 30 min, 2 hr, 3 hr, 4 hr, 5 hr, 6 hr, 24 hr), bath temperature (37.3 0 C), vessel temperature (37 "C), pump prime (60 seconds), pump purge
  • the media used was 900 g of SIFsp, pH 8.
  • DMSO DMSO (2.0 mL) was added to each test tube in the fraction collector to prevent the drug from recrystallizing once is cooled in the tubes. All capsules initially floated even though they were inserted into capsule sinkers purchased from Varian. The capsules sank before 15 minutes of mixing.
  • Vessel 3 Samples from Vessels 1 and 3 bounced out of the pill droppers.
  • the sample for vessel 1 was added to the open pill dropper in time to make it into the vessel.
  • the sample for vessel 3 did not get added to the pill dropper before it closed and a separate opening was opened and it was dropped through that opening by hand.
  • Vessel 3 was started approximately 20-30 seconds late.
  • Examples 144-146 These Examples describe the dissolution of carbamazepine and carbamazepine solid dispersions by evaluating the samples described in Examples 91-99. SIFsp, pH 6.8 media preparation was described in the Materials and Methods section.
  • FIG. 1 shows carbamazepine and carbamazepine solid dispersions dissolution profiles.
  • Examples 147-149 These Examples describe the dissolution of glyburide and glyburide solid dispersions by evaluating the samples described in Examples 109-117. SIFsp, pH 6.8 media preparation was described in the Materials and Methods section. Samples were transferred into gelatin capsules (Capsugel, size OCS, Lot #624282) using a manual single capsule filler. The weight of each capsule and the amount of sample added are found in Table 18 (Capsule Description).
  • FIG. 2 shows glyburide and glyburide solid dispersions dissolution profiles.
  • FIG. 3 shows the dissolution profile of the glyburide solid dispersions.
  • FIG. 4 is the dissolution profile of three different CMCAB/glyburide solid dispersions (see Tables 19 and 20 for the description of each sample). Both samples with lower dissolution rates (EX000039-036-2 and -036-3) were solid dispersions with approximately 2.7 % crystallinity. It is possible that the increase in crystallinity caused the decrease in release rate and total amount of glyburide released into the media. Other process factors could have also played a role in the reduced release rate.
  • FIG. 5 contains the dissolution profiles of two HPMCAS/glyburide solid dispersions compared with the best performing CMCAB/glyburide solid dispersion and glyburide (see Tables 19 and 20 for the description of each sample). It is obvious from these results that the release rate of glyburide can be modified within a group of solid dispersions using the same polymeric carrier and that CMCAB can perform equally as well as HPMCAS in certain systems.
  • FIG. 6 The mass of glyburide released from a CMCAB/glyburide solid dispersion and a HPMCAS/glyburide solid dispersion is presented in FIG. 6.
  • CMCAB/glyburide solid dispersion (EX000039-036-1 ) performed better than the HPMCAS/glyburide solid dispersion (EX000039-036-4).
  • HPMCAS/glyburide solid dispersion (EX000039-036-5) outperforms both of these samples as can be seen in FIG. 5.
  • the 036-5 sample had a lower drug loading than the 036- 1 or 036-4 samples.
  • Griseofulvin release from a CMCAB solid dispersion showed a more controlled release than the rapid release of griseofulvin from PVP solid dispersions, HPMCAS solid dispersion, or unmodified griseofulvin (FIGs. 7, 10- 13). However, the total amount of griseofulvin released was not improved by the formation of the solid dispersions.
  • Examples 162-164 These Examples describe the impact of surfactant additives on the dissolution profiles of griseofulvin/CMCAB solid dispersions.
  • Preparations of the griseofulvin/CMCAB, griseofulvin/CMCAB/Tween 80, and griseofulvin/CMCAB/SDS solid dispersions evaluated in this example are described in Examples 135-138.
  • SIFsp, pH 6.8 media preparation was described in the Materials and Methods section.
  • the surfactant was added prior to the co-evaporation process, and changed the near zero-order release profile of a griseofulvin/CMCAB solid dispersion into a much faster release profile.
  • the addition of the surfactant to the griseofulvin/CMCAB solid dispersion also increased the total amount of the drug released in the system when compared to the drug alone and the griseofulvin/CMCAB solid dispersion without the surfactant.
  • This Example describes the impact of polymer and plasticizer levels.
  • the use of a plasticizer that is mutually compatible with both the drug substance and the polymeric support in a solid dispersion may reduce the level of crystallinity of the drug substance trapped in the solid dispersion by generating a system of compatible ingredients and reducing the likelihood of drug substance "pooling" that would ultimately result in crystallization of the drug substance within the solid dispersion.
  • the impact of various plasticizers and plasticizer levels on the % crystallinity of a solid dispersion containing C-A-P or CMCAB and ibuprofen was investigated.
  • FIG. 15 shows the impact of TPGS on % crystallinity of ibuprofen/CMCAB solid dispersions (D-Optimal Mixture DOE Results).
  • This Example describes the impact of sample preparation method.
  • Solid dispersions were prepared using co-precipitation methods in which the drug, enteric polymer, and additives were dissolved in acetone then precipitated by adding the mixture to water. The poor water solubility of the drug results in the drug co-precipitating with the enteric cellulosic to produce a solid dispersion.
  • Evaluated here are a series of strategies for preparing solid dispersions, including co-precipitation (flake ppt'n and powder ppt'n), co-evaporation, and spray drying and determined the impact the various methods had on the % crystallinity of the solid dispersion, as indicated in Table 25, which shows the impact of method of preparation on % crystallinity of solid dispersions.
  • This Example describes the impact of process parameters (Temperature). Specifically, the impact of drying temperatures between 40 and 100 0 C on the % crystallinity of solid dispersions was evaluated. Increased drying temperatures or processing temperatures can reduce the crystallinity of a solid dispersion prepared by co-precipitation, as indicated in Table 26, which shows the impact of drying temperature on % crystallinity of ibuprofen/C-A-P solid dispersions.
  • Example # Amount Amount Description Amount Amount Wt % GIy 1 Wt % DMSO 2 % Xity 3 g g niL niL

Abstract

Cette invention concerne des compositions pharmaceutiques renfermant des esters de carboxyalkylcellulose utilisées pour administrer des substances pharmaceutiquement actives présentant une faible solubilité dans un milieu tel que l'eau, un tampon aqueux acide, un tampon aqueux neutre ou un tampon aqueux basique. Cette invention concerne également des procédés de production de compositions pharmaceutiques et des méthodes d'administration de ces compositions.
EP06827500A 2005-11-04 2006-11-03 Esters de carboxyalkylcellulose pour administration d'agents pharmaceutiquement actifs peu solubles Withdrawn EP1942872A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73349505P 2005-11-04 2005-11-04
PCT/US2006/043077 WO2007056205A2 (fr) 2005-11-04 2006-11-03 Esters de carboxyalkylcellulose pour administration d'agents pharmaceutiquement actifs peu solubles

Publications (1)

Publication Number Publication Date
EP1942872A2 true EP1942872A2 (fr) 2008-07-16

Family

ID=37888117

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06827500A Withdrawn EP1942872A2 (fr) 2005-11-04 2006-11-03 Esters de carboxyalkylcellulose pour administration d'agents pharmaceutiquement actifs peu solubles

Country Status (5)

Country Link
US (1) US20070178152A1 (fr)
EP (1) EP1942872A2 (fr)
JP (1) JP2009514884A (fr)
CN (1) CN101299993A (fr)
WO (1) WO2007056205A2 (fr)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9427419B2 (en) 2005-09-12 2016-08-30 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (DMSO)
AU2006291134C1 (en) 2005-09-12 2013-08-15 Abela Pharmaceuticals, Inc. Systems for removing dimethyl sulfoxide (DMSO) or related compounds, or odors associated with same
WO2007033180A1 (fr) 2005-09-12 2007-03-22 Abela Pharmaceuticals, Inc. Matieres facilitant l'administration du sulfoxyde de dimethyle (dmso) et de composes du meme type
WO2009076553A1 (fr) * 2007-12-11 2009-06-18 Monroe Stephen H Composition de solution tampon aqueuse pour le traitement d'un environnement cellulaire et de canaux ioniques et procédés d'utilisation de celle-ci
NZ588460A (en) * 2008-04-15 2012-07-27 Schering Corp Oral pharmaceutical compositions in a solid dispersion comprising preferably posaconazole and hpmcas
AU2009236290A1 (en) * 2008-04-15 2009-10-22 Merck Sharp & Dohme Corp. Oral pharmaceutical compositions in a solid dispersion comprising preferably posaconazole and HPMCAS
CN101606908B (zh) * 2009-07-10 2013-11-20 广州华农大实验兽药有限公司 癸氧喹酯可溶性粉及其制备方法
KR20120093993A (ko) 2009-10-30 2012-08-23 아벨라 파마슈티칼스, 인코포레이티드 퇴행성 관절염 치료를 위한 디메틸설폭사이드 및 메틸설포닐메탄 제제
CN101817057B (zh) * 2010-04-21 2012-01-11 沈阳汇亚通铸造材料有限责任公司 一种铸造用自硬砂慢型固化剂及砂型的制备
BR112012031667A2 (pt) * 2010-06-14 2016-11-08 Dow Global Technologies Llc composição
US9084727B2 (en) 2011-05-10 2015-07-21 Bend Research, Inc. Methods and compositions for maintaining active agents in intra-articular spaces
EP2736486B1 (fr) * 2011-07-28 2019-03-06 3M Innovative Properties Company Compositions cicatrisantes et leur méthode d'utilisation
WO2013106433A1 (fr) * 2012-01-09 2013-07-18 Virginia Tech Intellectual Properties, Inc. Dérivés de cellulose pour inhiber la cristallisation de médicaments médiocrement solubles dans l'eau
WO2013181174A2 (fr) 2012-05-31 2013-12-05 Merck Sharp & Dohme Corp. Formulations pharmaceutiques solides d'un antagoniste de récepteur d'orexine
KR102345084B1 (ko) * 2013-11-08 2021-12-29 액티버스 파마 컴퍼니 리미티드 매크롤라이드계 항균제의 나노미립자를 함유하는 수성 현탁액제
JP6680297B2 (ja) * 2015-04-28 2020-04-15 アステラス製薬株式会社 経口投与用医薬組成物
US20160331689A1 (en) * 2015-05-12 2016-11-17 SE Tylose USA, Inc. Aqueous enteric coating composition
EP3590505A1 (fr) 2015-08-08 2020-01-08 Alfred E. Tiefenbacher (GmbH & Co. KG) Formulation gastro-résistante contenant du posaconazole
CN107028931A (zh) 2016-02-04 2017-08-11 上海宣泰医药科技有限公司 一种紫杉醇药物组合物及其药物制剂、制备方法和用途
WO2017151455A1 (fr) * 2016-03-02 2017-09-08 Purdue Research Foundation Matériaux nanocristallins de cellulose fonctionnalisés et procédés de préparation
EP3705118B1 (fr) * 2017-10-31 2024-02-14 Samyang Holdings Corporation Composition pour forme posologique solide orale présentant une désintégration améliorée et son procédé de préparation
CN112110816A (zh) * 2020-09-02 2020-12-22 山东吉青化工有限公司 一种多元醇脂肪酸酯增塑剂的制备方法
EP4346776A1 (fr) * 2021-05-24 2024-04-10 Eastman Chemical Company Compositions pharmaceutiques contenant des solutions solides amorphes stables et des dispersions
CN115089555B (zh) * 2022-07-25 2023-11-07 上海药坦药物研究开发有限公司 一种卡马西平固体片剂及其制备方法
CN116285012A (zh) * 2023-03-15 2023-06-23 广东云启科技研究有限公司 纤维素基抗菌材料及其制备方法和应用

Family Cites Families (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3789117A (en) * 1971-07-19 1974-01-29 Freunt Ind Co Ltd Process for the preparation of enteric medicaments
FR2585246A1 (fr) * 1985-07-26 1987-01-30 Cortial Procede d'obtention de formes pharmaceutiques solides a liberation prolongee
US4775535A (en) * 1986-04-04 1988-10-04 Hans Lowey Method of preparing controlled long-acting pharmaceutical formulations in unit dosage form having uniform and comparable bioavailability characteristics
GB8613689D0 (en) * 1986-06-05 1986-07-09 Euro Celtique Sa Pharmaceutical composition
WO1995022962A1 (fr) * 1994-02-23 1995-08-31 Bm Research A/S Composition a liberation regulee
GB9405304D0 (en) * 1994-03-16 1994-04-27 Scherer Ltd R P Delivery systems for hydrophobic drugs
US5668273A (en) * 1996-01-29 1997-09-16 Eastman Chemical Company Carboxyalkyl cellulose esters
US5792856A (en) * 1996-01-29 1998-08-11 Allen; John Michael Process for preparing carboxyalkyl cellulose esters
US5781865A (en) * 1996-05-20 1998-07-14 Scientific Research Corporation PCS cell site system for allowing a plurality of PCS providers to share cell site antennas
IT1284604B1 (it) * 1996-09-27 1998-05-21 Roberto Valducci Composizioni farmaceutiche a rilascio controllato per somministrazione orale contenenti nifedipina come sostanza attiva
IL119627A (en) * 1996-11-17 2002-03-10 Yissum Res Dev Co PHARMACEUTICAL PREPARATIONS FOR THE CONTROLLED-RELEASE OF AN ACTIVE AGENT COMPRISING AT LEAST ONE β-LACTAM ANTIBIOTIC AGENT
US5968895A (en) * 1996-12-11 1999-10-19 Praecis Pharmaceuticals, Inc. Pharmaceutical formulations for sustained drug delivery
IT1289160B1 (it) * 1997-01-08 1998-09-29 Jagotec Ag Compressa farmaceutica completamente rivestita per il rilascio controllato di principi attivi che presentano problemi di
US6197787B1 (en) * 1997-03-03 2001-03-06 Sanofi-Synthelabo Pharmaceutical formulations containing poorly soluble drug substances
US6210710B1 (en) * 1997-04-28 2001-04-03 Hercules Incorporated Sustained release polymer blend for pharmaceutical applications
US5994530A (en) * 1998-06-25 1999-11-30 Eastman Chemical Corporation Carboxyalkyl cellulose esters for use in aqueous pigment dispersions
ES2306646T3 (es) * 1999-02-09 2008-11-16 Pfizer Products Inc. Composiciones de farmacos basicos con biodisponibilidad incrementada.
US6706283B1 (en) * 1999-02-10 2004-03-16 Pfizer Inc Controlled release by extrusion of solid amorphous dispersions of drugs
US6500459B1 (en) * 1999-07-21 2002-12-31 Harinderpal Chhabra Controlled onset and sustained release dosage forms and the preparation thereof
SE9903236D0 (sv) * 1999-09-10 1999-09-10 Astra Ab Method to obtain microparticles
SE0100823D0 (sv) * 2001-03-09 2001-03-09 Astrazeneca Ab Method I to obtain microparticles
SI1225874T1 (sl) * 1999-09-24 2006-06-30 Janssen Pharmaceutica Nv Protivirusne trdne disperzije
WO2001047498A2 (fr) * 1999-12-23 2001-07-05 Pfizer Products Inc. Forme de dosage d'un medicament stratifie entraine par un hydrogel
EP1120109A3 (fr) * 2000-01-24 2002-07-10 Pfizer Products Inc. Formes posologiques solides à désintégration et dissolution rapide
AR028253A1 (es) * 2000-03-16 2003-04-30 Pfizer Prod Inc Inhibidores de la glucogeno fosforilasa
DE60118742D1 (de) * 2000-07-07 2006-05-24 Kemestrie Inc Arzneistoffabgabesystem für wasser schwerlösliche arzneimittel
US7115279B2 (en) * 2000-08-03 2006-10-03 Curatolo William J Pharmaceutical compositions of cholesteryl ester transfer protein inhibitors
US6720003B2 (en) * 2001-02-16 2004-04-13 Andrx Corporation Serotonin reuptake inhibitor formulations
US6610326B2 (en) * 2001-02-16 2003-08-26 Andrx Corporation Divalproex sodium tablets
EP1404300B1 (fr) * 2001-06-22 2009-09-30 Pfizer Products Inc. Compositions pharmaceutiques de dispersions de medicaments et de polymeres neutres
WO2003024429A1 (fr) * 2001-09-21 2003-03-27 Egalet A/S Systeme de liberation a base de polymere
EP1429744A1 (fr) * 2001-09-21 2004-06-23 Egalet A/S Systeme a liberation de polymere de morphine
IN192180B (fr) * 2001-09-28 2004-03-06 Ranbaxy Lab
ES2292848T3 (es) * 2001-10-12 2008-03-16 Elan Pharma International Limited Composiciones que tienen una combinacion de caracteristicas de liberacion inmediata y liberacion controlada.
US20030091630A1 (en) * 2001-10-25 2003-05-15 Jenny Louie-Helm Formulation of an erodible, gastric retentive oral dosage form using in vitro disintegration test data
RU2278659C9 (ru) * 2002-02-01 2007-02-20 Пфайзер Продактс Инк. Способ получения гомогенных, высушенных распылением твердых аморфных дисперсий лекарственного средства с использованием модифицированной установки сушки распылением
US20030185774A1 (en) * 2002-04-02 2003-10-02 Dobbs Suzanne Winegar Cosmetic coating composition comprising carboxyalkyl cellulose ester
US7803402B2 (en) * 2002-07-06 2010-09-28 Sanjeev Khandelwal Pharmaceutical preparations
KR20040011248A (ko) * 2002-07-30 2004-02-05 환인제약 주식회사 암로디핀의 고체분산체와 그 제조방법 및 그를 포함하는약학적 조성물
AU2003275345A1 (en) * 2002-10-01 2004-04-23 Eastman Chemical Company Use of carboxyalkyl cellulose esters, such as carboxymethyl cellulose acetate butyrate, to form aqueous dispersions of hydrophobic materials in water
US20060053669A1 (en) * 2004-09-16 2006-03-16 Edwards Systems Technology, Inc. Method and apparatus for indicating a configurable switch

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007056205A2 *

Also Published As

Publication number Publication date
US20070178152A1 (en) 2007-08-02
WO2007056205A2 (fr) 2007-05-18
JP2009514884A (ja) 2009-04-09
WO2007056205A3 (fr) 2007-10-25
CN101299993A (zh) 2008-11-05

Similar Documents

Publication Publication Date Title
US20070178152A1 (en) Carboxyalkylcellulose esters for administration of poorly soluble pharmaceutically active agents
US10744086B2 (en) Fast dissolving solid dosage form
AU2008303129B2 (en) Compositions comprising lipophilic active compounds and method for their preparation
KR101411100B1 (ko) 친지성 약물을 함유하는 나노캡슐을 포함하는 미소구체
US6509040B1 (en) Fast dispersing dosage forms essentially free of mammalian gelatin
US20060105045A1 (en) Cyclodextrin solubilizers for liquid and semi-solid formulations
JP2010525082A (ja) 固体投与形
WO2000044351A1 (fr) Formes posologiques a diffusion rapide exemptes de gelatine
EP1737430A1 (fr) Methode de preparation d'oltipraz de faible cristallinite ou d'oltipraz amorphe
WO2012116066A1 (fr) Formulation d'hydrogel à libération contrôlée
CA3013530A1 (fr) Preparations pharmaceutiques oromuqueuses a biodisponibilite elevee a base de cyclodextrine et de sucralose
EP1942869A2 (fr) Esters de cellulose carboxyalkyle destines a une administration prolongee de substances actives sur le plan pharmaceutique
US20080075785A1 (en) Controlled release hydrogel formulation
Rehman et al. Polysaccharide-based amorphous solid dispersions (ASDs) for improving solubility and bioavailability of drugs
US20200016080A1 (en) Controlled release pharmaceutical compositions
US20170216217A1 (en) Spherical microcapsules with enhanced oral bioavailability
Kumar et al. Solid dispersions: An approach to enhance solubility of poorly soluble drugs
WO2020117736A1 (fr) Acétate-succinates d'hydroxypropylméthylcellulose de masse moléculaire très élevée
Sharma et al. Fast Dissolving Tablets-A Review and Recent Advances in Manufacturing Technologies

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080327

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: DRESSMAN, JENNIFER, JOY

Inventor name: POSEY-DOWTY, JESSICA DEE

Inventor name: EDGAR, KEVIN, JOSEPH

Inventor name: KLEIN, SANDRA

Inventor name: SHELTON, MICHAEL, CHARLES

Inventor name: LINGERFELT, LARRY, RONNIE, JR.

Inventor name: KIRK, SHANE, KIPLEY

17Q First examination report despatched

Effective date: 20080901

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090113